Next Article in Journal
Epigenetic Heterogeneity of B-Cell Lymphoma: Chromatin Modifiers
Next Article in Special Issue
COBRA-Seq: Sensitive and Quantitative Methylome Profiling
Previous Article in Journal
Transcriptome Changes during the Life Cycle of the Red Sponge, Mycale phyllophila (Porifera, Demospongiae, Poecilosclerida)
Previous Article in Special Issue
Current and Future Prospects for Epigenetic Biomarkers of Substance Use Disorders
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Characterization of DNA Methylation in Circulating Tumor Cells

by
Constantin F. Pixberg
1,
Wolfgang A. Schulz
2,
Nikolas H. Stoecklein
1 and
Rui P. L. Neves
1,*
1
Department of General, Visceral and Pediatric Surgery, University Hospital and Medical Faculty of the Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany
2
Department of Urology, University Hospital and Medical Faculty of the Heinrich-Heine University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
*
Author to whom correspondence should be addressed.
Genes 2015, 6(4), 1053-1075; https://doi.org/10.3390/genes6041053
Submission received: 3 July 2015 / Revised: 9 October 2015 / Accepted: 14 October 2015 / Published: 21 October 2015
(This article belongs to the Special Issue Epigenetic Biomarkers)

Abstract

:
Epigenetics contributes to molecular mechanisms leading to tumor cell transformation and systemic progression of cancer. However, the dynamics of epigenetic remodeling during metastasis remains unexplored. In this context, circulating tumor cells (CTCs) might enable a direct insight into epigenetic mechanisms relevant for metastasis by providing direct access to systemic cancer. CTCs can be used as prognostic markers in cancer patients and are regarded as potential metastatic precursor cells. However, despite substantial technical progress, the detection and molecular characterization of CTCs remain challenging, in particular the analysis of DNA methylation. As recent studies have started to address the epigenetic state of CTCs, we discuss here the potential of such investigations to elucidate mechanisms of metastasis and to develop tumor biomarkers.

1. Introduction

Metastasis is a multi-step process involving tumor cell migration, intravasation, survival in the blood stream, extravasation at a secondary site, and colonization. Despite significant progress in understanding of this complex metastatic cascade, there is still a significant lack of knowledge on the underlying molecular mechanisms, which is obviously a major obstacle for the development of more effective therapies. The fact that tumor-related mortality is due to metastasis in 90% of the patients emphasizes its tremendous clinical importance [1].
Systemic disease is driven by cells that have successfully accomplished at least some steps of the metastatic cascade. Compared to their primary tumors, these cells have usually accumulated additional genetic and phenotypic alterations as a result of ongoing genetic instability and the different selective pressures experienced in the various micro-environments [2]. In this respect, tumor cells that have left the primary tumor or metastases and that can be found in the circulation of patients (circulating tumor cells, CTCs) are an important source of information about this process. In addition to their heuristic value for understanding fundamental aspects of tumor progression, these cells have now been proven to be clinically relevant, and their molecular characterization is acknowledged as an opportunity for real-time monitoring of the disease progression and of the molecular evolution of metastatic systemic disease [3,4].
Another class of relevant blood-based biomarkers are cell-free nucleic acids, in particular DNA (cell-free DNA, cfDNA), which are found at increased concentrations in cancer patients. Different genetic and epigenetic analyses have already demonstrated the clinical potential of cfDNA [5]. cfDNA is thought to originate from apoptotic and necrotic cancer cells from the primary tumor site, CTCs, and distant metastases [6]. As cfDNA represents a source of tumor genetic material, its analysis can serve as a convenient alternative or a complement to CTCs-based assays. However, since the origin of cfDNA can be very heterogeneous and cannot be traced to specific tumor cell populations, analysis of CTCs is preferable especially when exploring mechanisms of metastasis. Despite its potential, cfDNA is outside the scope of our review focusing on CTCs; reviews on cfDNA can be found in [5,6].
So far, molecular analyses of CTCs have rather focused on their genomes, transcriptomes and proteomes. In contrast, the epigenetics of CTCs remains a largely unexplored field with great potential. Considering the accumulated evidence for the role of epigenetics, especially DNA methylation, in several cellular mechanisms of metastasis, the low number of studies addressing the epigenetics of CTCs is surprising. However, the few existing studies on DNA methylation of CTCs already provide interesting initial evidence for epigenetic remodeling in these cells and for the clinical potential of CTC-epigenetics. They will therefore certainly provide a basis for future investigations. In this review we discuss the potential of CTCs and DNA methylation analyses for elucidating the metastatic cascade. We then review in detail the existing literature on DNA methylation in CTCs, discuss the major technical challenges for their epigenetic analysis, and provide our perspective on the potential contained in a deeper epigenetic characterization of CTCs.

2. The Utility of CTCs as Tumor Biomarkers

Today, CTCs are investigated not only for their utility as biomarkers, but also because they provide an opportunity to directly access tumor material via a simple blood draw. The CellSearch® system was the first semi-automated system designed specifically for the enrichment and enumeration of CTCs [7], and it is still the only system approved by the Federal Drug Agency (FDA) for that purpose. The clinical utility of CTCs detected by the CellSearch® system is now widely recognized in different types of tumors including breast, prostate and colorectal carcinomas. In these cancers, CTC-counts above defined thresholds have prognostic impact, correlating with reduced progression-free and overall survival of the patients [8,9,10,11,12,13,14,15]. For breast cancer, several studies have moreover provided good evidence that CTCs might serve as markers for monitoring disease response to therapy at different disease stages and in diverse clinical settings. In the neoadjuvant setting, persistence of CTCs after therapy identifies a group of patients with significant increased risk for early metastatic relapse and reduced overall survival [16,17]. In the adjuvant setting, the large SUCCESS trial demonstrated that the detection of CTCs in 30 mL blood before therapy was significantly associated with poor disease-free survival, distant disease-free survival, breast cancer-specific survival, and overall survival [18]. In metastatic breast cancers, patients with five or more CTCs per 7.5 mL blood before initiation of therapy have worse progression-free survival and overall survival [8]. Also, in metastatic breast disease, CTC enumeration can help to monitor disease response to treatment [19,20]. In particular, the development or persistence of high CTC-counts during the course of therapy is a strong indicator of a worse outcome, shorter time to progression and overall survival [9,11,21]. Importantly, a multinational, randomized phase III trial involving 711 patients with metastatic castration-resistant prostate cancer (mCRPC), demonstrated the utility of CTC enumeration also as a clinical trial end point [22]. In particular, the study showed that patients with less than five CTCs in 7.5 mL blood at week 12 constituted a low-risk group with a significantly longer overall survival. In this study CTC counts combined with lactase dehydrogenase (LDL) levels (both measured at week 12 of treatment) satisfied all four Prentice criteria [23] as efficacy-response surrogate for overall survival, indicating that CTC enumeration at week 12 could be used as a valid end point criterion. Thus, CTC enumeration might allow obtaining information from clinical studies within much shorter time frames.
Despite the solidly documented prognostic value of CTCs, the predictive value of CTC enumeration per se could not yet been shown in prospective clinical trials [21]. In a manner, this inability underlines the relevance of going beyond enumeration to explore the molecular characteristics of CTCs. In fact, CTCs have been already investigated for the targets of molecular therapies. Notably, different studies revealed molecular differences in HER2 [24,25,26], ER and PR status between breast cancer CTCs and their matched primary tumors, and likewise in their gene expression profiles [27,28]. Strengthening the relevance of molecular characterization of CTCs as a complement to the analysis of primary tumors, Meng and colleagues verified that three out of four breast cancer patients with HER2-positive CTCs benefited from HER2-targeted therapy, even though the primary tumors were HER2-negative [24]. More recently, a prospective clinical study has demonstrated that the presence of androgen receptor splice variant 7 (AR-V7) mRNA in CTCs of patients with metastatic castration-resistant prostate cancer (mCRPC) was consistently associated with resistance to two compounds targeting the AR-response (enzalutamide and abiraterone) [29]. In this study involving 62 CTC-positive mCRPC patients, AR-V7 could be detected in baseline samples of 18 patients none of whom benefited from either therapy. In addition, positivity for AR-V7 in secondary end points was statistically significant associated with shorter overall survival and other outcome parameters. Further prospective studies involving larger cohorts are underway to validate the important clinical implications of this study.
Importantly, besides their clinical relevance, CTCs constitute also an important source of material to address the metastatic cascade. Recently, evidence was obtained that patient-derived CTCs comprise indeed tumor initiating cells by injecting them directly into immunocompromised mice [30]. In addition, among patient CTCs, a subset of EpCAM+/CD44+/CD47+/MET+ CTCs had the potential to seed metastases in bone, lung and liver [31]. Further, one other independent study has also demonstrated that EpCAM-negative cells expressing HER2, EGFR, HPSE, and NOTCH1, isolated from patient-derived CTC cell lines have the potential to form metastases in lung and brain of mice [32].
The potential of exploring CTCs for a better understanding of the metastatic cascade and for clinical applications is hampered by technical difficulties. Even in metastatic patients, CTCs are extremely rare (their frequency can be as low as one CTC per 105–107 mononuclear cells) [33], which imposes limitations for their detection and detailed characterization. One factor contributing to the low number of CTCs is the commonly used low blood volume of 7.5–10 mL. One approach to circumvent this limitation employs diagnostic leukapheresis (DLA), which allows screening of up to 2.5 L of peripheral blood for CTCs and substantially enhances the CTC detection frequency [34]. Other limitations are associated with the enrichment and detection of CTCs. To address these, several methodologies and devices have been developed that exploit phenotypical (e.g., expression of surface markers) and/or morphological (e.g., size, density, deformability) differences between CTCs and normal blood cells [35,36]. The CellSearch® system used in the majority of clinical CTC studies relies on EpCAM-based immunomagnetic enrichment and detection of nucleated, cytokeratin 8/18/19-positive, but CD45-negative cells [7]. The specificity of this system was demonstrated by the finding that virtually all CTCs enriched by CellSearch® display aCGH profiles with typical structural chromosomal changes expected for the respective tumors [37,38]. Many other systems also rely on EpCAM for the enrichment of CTCs [36], such as the Isoflux™ system based on microfluidics [39], the MagSweper [40], the CTC-iChip, which combines EpCAM-based enrichment and morphological criteria for the identification of CTCs [41], or the CellCollector®, a functionalized wire inserted into the cubital vein for 30 min for in vivo capturing of CTCs [42]. For a comprehensive review on methods available for CTC enrichment, please see Yap et al., 2014 [36].
A critically discussed issue with all EpCAM-based capture strategies is that EpCAM on carcinoma cells, specifically those in the circulation, may become downregulated [43]. Evidence from metastasizing xenograft models suggests that this downregulation may happen rapidly, within the first hour of circulation in blood [44]. There are several explanations for lack of EpCAM expression on CTCs [45], including epithelial-to-mesenchymal transition (EMT), which is discussed as an important mechanism for dissemination and metastasis. In fact, subsets of CTCs with a mesenchymal-like phenotype can be observed in the circulation [46] and seem to accrue with advanced disease stages [47,48]. This phenotypical heterogeneity among CTCs imposes obvious limitations on any technique for their enrichment and identification [49,50]. To circumvent the dependency on EpCAM, other methods have been developed for less restrictive enrichment of CTCs [36,51]. These include filter systems that rely on the larger size of CTCs like the Rarecells® device [52], the ScreenCell® system [53], the CellSieve™ system [54], or systems that rely on functional properties of CTCs like the Vita-Assay™, which is based on the ability of CTC to adhere to and invade a matrix membrane [55]. Following enrichment, the distinction of CTCs from co-enriched normal blood cells again requires tumor-specific markers, which are typically identified by immunostaining or in situ hybridization. Like the biological markers used for enrichment, the markers used for the identification of CTCs are not undisputed and need more validation. To achieve standardization of technologies, markers and assays for CTC detection and analysis, more than 30 European academic and private institutions have joined efforts in an Innovative Medicines Initiative (IMI) called CANCER-ID (started in January 2015).
In conclusion, CTCs seem to be relevant for systemic progression and can provide access to the characteristics of the systemic disease in the sense of a “liquid biopsy” for diagnosis, prognosis and eventually guidance of therapeutic decisions. Furthermore, their direct molecular characterization may contribute to a better understanding of the metastatic process.

3. Biological and Clinical Relevance of DNA Methylation in Cancer

DNA methylation acts as a major epigenetic mechanism within the complex network of mechanisms regulating gene expression [56]. The chemically stable addition of a methyl group to cytosines interferes with the capacity of several proteins to bind DNA [57]. Methylation in CpG-rich gene promoters serves as a recognition signal for proteins mediating transcriptional repression and constitutes a mechanism for stable gene silencing, whereas methylation of intragenic regions usually correlates with increased gene transcription [58,59]. For the regulation of tissue-specific expression, DNA methylation levels at enhancer sequences appear to be crucial. Methylation in repetitive transposable elements plays an important role in their silencing and contributes to global genomic stability [60]. Physiologically, methylation patterns are established during early embryogenesis and contribute to lineage commitment and cellular differentiation. These patterns are faithfully maintained during DNA replication by specialized DNA (cytosine-5-)-methyltransferases (DNMTs), but under several pathological conditions global changes in the epigenetic landscape take place [61,62]. In tumours, global methylation levels are often diminished (global hypomethylation) whereas simultaneously methylation is gained at specific loci (local hypermethylation). In various cancers, hypomethylation is observed at normally methylated retroelements [60,63], while hundreds to thousands of individual genomic loci become hyper- or hypomethylated compared to matched normal tissues [64].
Interestingly, methylation patterns established during tumorigenic cellular transformation and subsequent tumor progression are not random, suggesting that the rearrangements of the methylation landscape during carcinogenesis are orchestrated in a tumor-specific manner [65]. Examples of this orchestrated DNA methylation remodeling can be found in several genes involved in EMT [66], a process thought to drive tumor cell dissemination [67], acquisition of stem cell properties [68], and to be crucial for the fate of CTCs [69]. These genes comprise among others those encoding E-cadherin (CDH1), TWIST, Vimentin (VIM), N-cadherin (CDH2) and the miR-200 family of miRNAs [70,71,72,73,74]. Notably, changes in DNA methylation during the induction of EMT in MDCK cells by TGFβ and its reversion (MET) are dynamic [66,75]. Stable alterations in DNA methylation can therefore serve as a biomarker for cancer, but some changes in DNA methylation might also be used to follow the dynamics of cellular phenotypic plasticity as it occurs during metastasis.
Another example of a group of epigenetically regulated genes relevant for metastasis are “metastasis suppressor genes”. These are defined as genes that do not substantially affect tumor growth at the primary site, but must be inactivated to permit metastasis [76]. Typically, inactivation of these genes occurs not by mutations, but by epigenetic mechanisms, which may be dynamic during tumor progression. For instance, mice lacking the Epb41l3 gene do not develop cancers spontaneously, but metastasis of autochthonous prostate cancer is enhanced [77]. This gene is downregulated by hypermethylation in several human carcinomas, including prostate cancer [78]. It will be highly interesting to find out whether methylation of such genes is accentuated also in CTCs.
The importance of DNA methylation in cancer has led to the development and testing of several drugs that interfere with the regulation of this epigenetic modification. The general idea of epigenetic therapies is the re-establishment of more normal patterns of gene expression which had become deregulated in cancer cells. Azacitidine (Vidaza) and its deoxy derivative 5-aza-2'-deoxycytidine (Decitabine) (both FDA-cleared epigenetic drugs) are nucleoside analogues that are incorporated into DNA during replication instead of cytidine and, by inactivating and depleting DNMTs, lead to global hypomethylation of the genome. These drugs, alone and in combination with other chemotherapeutic agents, have proven their benefits especially in patients with hematologic tumors (e.g., myelodysplastic syndrome [79,80], and acute myeloid leukemia [81,82,83]). In solid tumors, their application is more controversial [84,85]. However, in the management of some refractory advanced solid tumours, low-dose Decitabine-based chemoimmunotherapy has shown promise [86].
The chemical stability of cytosine methylation, the biological stability of methylation patterns through cell division, and the tumor-specific re-organization of the methylation landscape, taken together, make DNA methylation an attractive source of tumor biomarkers. One good example is the promoter methylation of the DNA repair gene MGMT coding for O-6-methylguanine-DNA methyltransferase. Glioblastomas [87] and stage IV melanomas [88] with MGMT methylation respond better to treatment with alkylating agents (temozolomide and dacarbazine), and MGMT methylation is an independent variable associated with disease-free [88] and overall survival [87]. In fact, MGMT methylation is now routinely used for selection of glioblastoma patients for temozolomide treatment [89]. In colorectal cancer as well, MGMT hypermethylation is significantly associated with a favourable clinical response to dacarbazine [90]. Several other methylation events have also been identified as prognostic markers. For instance, methylation of the VIM gene encoding the EMT indicator vimentin is associated with a markedly decreased survival in breast cancer [72]. Contrarily, it is also suggested as a biomarker of favorable prognosis in pancreatic cancer [91] and uterine cervical squamous cell carcinoma [92]. Hypermethylation of VIM can be used for detection of colorectal cancer using stool samples [93] and of urothelial carcinoma using urine [94,95].
Interestingly, in most cancers, more genes are affected by alterations in DNA methylation than by somatic genetic mutations [96], offering a much wider range of targets to design assays. Moreover, in contrast to genetic mutations, DNA methylation can be reversed and tumor regression might also involve the reversal of such biomarks. The dynamics of epigenetic biomarkers might therefore provide a good tool for monitoring disease progression and response to treatment.
Several robust technologies are now available for analyzing DNA methylation in tumor material and body fluids. The choice between them depends on the scope, resolution, available biological material, and involved labor and costs [97,98]. The methods range from locus-specific, through large-scale analysis, to whole methylome approaches. Some of the methods allow precise determination of the methylation status at individual CG-dinucleotides, while others provide only estimates of methylation levels across defined genomic regions or the entire genome. Most current techniques for high-resolution methylation analysis require treatment of DNA with sodium bisulfite, which converts unmethylated cytosine to uracil, whereas methylcytosine remains unaffected. Subsequent to conversion, different methods can be used to discriminate between uracil and cytosine nucleotides, and to infer the presence of unmethylated or methylated cytosines in the original sequence (Figure 1).
Figure 1. Bisulfite conversion of DNA and methods used for high resolution mapping of methylcytosines. (Upper panel) DNA conversion upon treatment with bisulfite. Bisulfite reacts with non-methylated cytosine residues converting them to uracils by initial sulfonation, hydrolytic deamination and final desulfonation. Methylcytosines essentially do not react with bisulfite. After conversion, uracil residues indicate non-methylated cytosines, while cytosines indicate methylcytosines in the original DNA sequence; (Lower panel) Converted DNA can be analyzed by multiple techniques discriminating between cytosines and uracils to detect or precisely map methylcytosines. These include clone sequencing (Clone Seq.), pyrosequencing (Pyro Seq.), MALDI-TOF mass spectroscopy (Mass Spec.), bead arrays, bisulfite padlock probes (BSPP), solution hybrid selection (SHS), reduced-representation bisulfite sequencing (RRBS), and whole genome sequencing (MethylC-Seq). These methods differ in the size of the target sequence, the number of CpGs covered, as well as in their cost.
Figure 1. Bisulfite conversion of DNA and methods used for high resolution mapping of methylcytosines. (Upper panel) DNA conversion upon treatment with bisulfite. Bisulfite reacts with non-methylated cytosine residues converting them to uracils by initial sulfonation, hydrolytic deamination and final desulfonation. Methylcytosines essentially do not react with bisulfite. After conversion, uracil residues indicate non-methylated cytosines, while cytosines indicate methylcytosines in the original DNA sequence; (Lower panel) Converted DNA can be analyzed by multiple techniques discriminating between cytosines and uracils to detect or precisely map methylcytosines. These include clone sequencing (Clone Seq.), pyrosequencing (Pyro Seq.), MALDI-TOF mass spectroscopy (Mass Spec.), bead arrays, bisulfite padlock probes (BSPP), solution hybrid selection (SHS), reduced-representation bisulfite sequencing (RRBS), and whole genome sequencing (MethylC-Seq). These methods differ in the size of the target sequence, the number of CpGs covered, as well as in their cost.
Genes 06 01053 g001
Standard Sanger sequencing after cloning [99,100] or the more quantitative pyro-sequencing technique [101] provide single CpG resolution across a few hundred bps for individual loci. A matrix-assisted laser desorption/ionization time of flight (MALDI-TOF) mass spectrometry-based technique has the advantage of easy automatization [102,103]. Several bisulfite sequencing techniques are available for intermediate scale analyses of multiple pre-selected CpGs. They rely on the capture of target regions by padlock probes [104,105,106] or solution hybrid selection [107,108], and can be coupled to next generation sequencing to achieve high throughput. Another widely used technique at intermediate scale is hybridization to methylation bead arrays covering approximately 500,000 CpG sites across the genome [109,110]. Comprehensive, but expensive whole-genome bisulfite sequencing [111,112] and the less costly reduced representation bisulfite sequencing (RRBS) [113,114] are commonly used for high resolution analysis of entire genomes (methylomes).
The application of established epigenetic methods to rare cell populations, such as CTCs, remains very challenging, especially at the single-cell level. For various genetic and genomic analyses, DNA from single cells can be reliably amplified. However, DNA methylation patterns are not maintained during DNA amplification, and analysis of methylation therefore requires bisulfite treatment of DNA prior to amplification. This poses a technical challenge, especially for samples composed of few cells as a minute amount of DNA needs to be carried through several treatment and purification steps before the actual analyses. In addition, bisulfite treatment causes random DNA fragmentation, which can be a limitation for subsequent analyses. Nevertheless, some bisulfite-based protocols have already been successfully applied to few or even single-cells by RRBS [115] or single-cell locus-specific bisulfite sequencing (SLBS) [116]. These studies have addressed DNA methylation changes during embryonic development illustrating the potential of these protocols for detecting inter- and intracellular methylation heterogeneity. Further development of these techniques will be needed for the systematic and robust analysis of DNA methylation which would be necessary to explore the epigenetic heterogeneity of tumor cells and CTCs at the single-cell level.

4. DNA Methylation in CTCs

The number of studies on methylation analysis of CTCs is so far small (Table 1). This is most likely explained by the combined technical challenges of CTC isolation and DNA methylation analyses on extremely rare cells.
In a series of three pioneering studies, Chimonidou and colleagues [117,118,119] investigated the methylation status of three tumor-associated genes in tumor cells from the blood of patients with breast cancer. The selected genes comprised Cystatin M6 (CST6), breast cancer metastasis suppressor 1 (BRMS1), and SRY (sex-determining region Y)-box 17 (SOX17). Cystatin M6 (E/M), an endogenous inhibitor of cathepsins B and L [120,121], is a postulated tumor suppressor in breast cancer [121,122]. Epigenetic silencing of CST6 had already been associated with unfavorable prognosis in operable breast cancer [123,124]. BRMS1 was described as a metastases-suppressing protein [125] whose expression is modulated by DNA methylation [126]. Its reduced expression was correlated with poor prognosis [127,128] and predicted shorter disease-free survival in breast cancer [129]. SOX17, controlled by DNA methylation, encodes a transcription factor with a potential tumor suppressor function by antagonizing WNT-signaling [130,131]. Its reduced expression was associated with tumor progression and poor prognosis in breast cancer [132], and with patient survival in melanoma [133]. Considering the evidence pointing to the biological and clinical importance of epigenetic remodeling in these genes, Chimonidou and colleagues investigated whether methylation of these genes could also be detected in breast CTCs and could yield prognostic information. In a first study [117], the authors enriched EpCAM-expressing cells from the blood of patients with early and metastatic breast cancer or healthy controls by immunomagnetic capturing, and analyzed the methylation at the promoter regions of CST6, BRMS1, and SOX17 by methylation-specific PCR (MSP) categorizing the samples as positive/negative. The frequency of methylation at the three gene promoters was significantly higher in EpCAM-enriched cells from patients than in those from healthy donors [117]. Notably, the authors observed that the frequency of methylation at these genes tended to increase from patients with operable tumors to patients with metastatic disease [117].
Table 1. Technical details and key findings of publications addressing DNA methylation in CTCs.
Table 1. Technical details and key findings of publications addressing DNA methylation in CTCs.
ReferenceEntityCTC PreparationMethylation AnalysisKey Findings
EnrichmentDetection and IsolationLocusMethod
Chimonidou, Strati et al., 2011 [117]BC (Human)Density centrifugation + EpCAM-coated beads-CST6, BRMS1, SOX17MSPPositive correlation with presence and stage of disease.
Chimonidou, Strati et al., 2013 [118]BC (Human)Density centrifugation + EpCAM-coated beads-SOX17MSP (real-time)Strong correlation between methylation detected in CTCs and cfDNA.
Chimonidou, Kallergi et al., 2013 [119]BC (Human)Density centrifugationDetection of CK+ cells on cytospins by ICC and scratchingBRMS1MSPWeak correlation between methylation and protein expression. Discordant methylation in CTCs and patient-matched PTs.
Friedlander, Ngo et al., 2014 [134]mCRPC (Human)CAM-Genome wide (27000 CpG)Methylation arrayCorrelation between methylation in CTCs and non-matched PTs. Hypermethylation in apoptosis, angiogenesis, and VEGF pathway genes.
Ogunwobi, Pussyk et al., 2013 [135]HCC (Murine cell line)Implantation of tumor cells in mice, and establishment of CTC linesHGF, c-MetHRM and PyrosequencingHGF and c-MET overexpression in the CTC-lines correlated with hypomethylation of their promoters.
Notes: BC—breast cancer; mCRPC—metastatic castration-resistant prostate cancer; HCC—hepatocellular carcinoma; CAM—cell-adhesion matrix; MSP—methylation-specific PCR; HRM—high resolution melting; cfDNA—cell-free DNA; PTs—primary tumors.
Cell-free DNA (cfDNA) circulating in blood, too, has been proposed as a relevant source of tumor material and biomarkers [136], but its association with CTCs is still obscure. In a second study [118], Chimonidou and colleagues therefore explored the correlation between SOX17 promoter methylation in EpCAM-expressing cells enriched from blood and in cfDNA isolated from plasma of patient-matched samples. Similarly to their previous study, the authors assessed DNA methylation by MSP and stratified the samples as positive/negative. In total, the authors detected SOX17 promoter methylation in 4.3%, 34.5%, and 45.8% of EpCAM-enriched cell fractions, and in 2.0%, 34%, and 40.7% of cfDNA samples, respectively, from healthy donors, patients with operable tumors, and patients with metastatic tumors. In the group of operable breast cancers, the authors reported a significant (p = 0.008) correlation between methylation detected in EpCAM-enriched cells and matched cfDNA.
In a third study [119], the same group investigated the association between BRMS1 promoter methylation and the expression of its encoded protein in CTCs. For that, the authors analyzed the expression of BRMS1 protein and Cytokeratins 8, 18, and 19 by immunofluorescent staining on cytospin preparations of peripheral mononuclear cells (PBMNCs) enriched via Ficoll density gradient centrifugation from peripheral blood of patients with operable breast cancer. The intensity of the BRMS1 staining in CK-expressing cells was semi-quantitatively measured (negative/low/high), using the epithelial breast cancer MCF7 cell line and normal PBMCs as standards. To evaluate BRMS1 promoter methylation in the CK-positive cells, the authors scratched these from the cytospins, pooled the material, and extracted the DNA for bisulfite treatment and qualitative MSP. They observed a weak correlation between BRMS1 promoter methylation and lack of protein expression. CTCs lacking BRMS1 protein were detected in eight samples, but methylation of the BRMS1 promoter could be detected only in four of these. Instead, BRMS1 gene promoter methylation was detected in one sample without CTCs. A further interesting observation was a high discordance between the methylation statuses of BRMS1 in CK-positive cells and matched primary tumor samples.
Collectively, these pioneering studies demonstrate that tumor-associated methylation can indeed be detected in cell-fractions enriched for CTCs and provide a first glimpse into the dynamics of epigenetics during human tumor cell dissemination. However, limitations of these three studies were the use of unpurified CTC samples and of the MSP method, which does not provide a quantitative measure of DNA methylation or information on methylation patterns.
A different approach to explore epigenetic features in CTCs was taken by Friedlander and colleagues [134]. These authors enriched viable CTCs by their invasive phenotype from a patient with metastatic castration-resistant prostate cancer (mCRPC) before chemotherapy by taking advantage of the ability of tumor cells to invade a cell-adhesion matrix. This system for capturing CTCs (Vita-Assay™) had been previously described to allow efficient recovery of tumor cells that correlated with clinico-pathological parameters of patients with early stage breast cancer [137], and ovarian cancer [55]. Cells enriched by this technique can be released from the matrix and propagated in culture, or stained for precise enumeration and subsequent molecular characterization, e.g., by array comparative genomic hybridization [137,138]. The invasive cells enriched by this method from the mCRPC patient were then analyzed using a methylation array covering 27,000 CpG-sites [134]. Across all CpGs, the enriched cells presented an average methylation value comparable to that of primary mCRPC tumors, which was higher than in benign prostatic tissues. Notably, 86% of the 1361 loci reported as hypermethylated in mCRPC tumor samples in a previous study [139], were also methylated in the recovered invasive CTCs. The percentage of overlapping loci between these two studies increased to 95% if less stringent criteria were used to call for methylation in the invasive cells. Pathway analyses indicated that methylation in the invasive tumor cells occurred more frequently in genes associated with apoptosis, angiogenesis, and VEGF signaling. This exploratory genome-wide analysis has provided a first insight into global epigenetics of CTCs suggesting that some of the epigenetic programs associated with tumor progression detectable in primary tumors are maintained in CTCs.
In addition to the studies based on ex vivo analyses of patient-derived CTCs, interesting epigenetic CTC data were obtained from a syngeneic murine hepatocellular carcinoma (HCC) model. Ogunwobi et al. employed this approach to address the role and mechanisms regulating the expression of hepatocyte growth factor (HGF) in CTCs [135]. After orthotopic or subcutaneous implantation, the murine HCC cell-line BNL 1ME A.7R.1 gave rise to detectable tumors and CTCs. The authors established three cell lines from CTCs isolated from these mice for functional and molecular testing. The established CTC cell lines showed increased tumorigenicity and metastatic potential compared to their mother cell line and displayed a more mesenchymal phenotype (increased expression of vimentin, fibronectin, and collagen I, combined with decreased expression of E-cadherin). Concordantly, the hepatocyte growth factor (HGF) and its receptor, the proto-oncogene c-MET, were more strongly expressed in the CTC lines. Accordingly, as in many other cell types [140], HGF induced EMT features in the BNL 1ME A.7R.1 cell line. To explore the mechanisms regulating HGF and c-MET, the authors performed analyses of DNA methylation at the promoter of both respective genes using bisulfite-conversion and high resolution melting analysis, which allows an estimate of methylation across a given PCR amplicon. Indeed, overexpression of HGF and c-MET in the CTC lines was accompanied by a decrease of methylation at their promoters. For c-MET, albeit not for HGF, the results from the high resolution melting analysis were confirmed by the more reliable and quantitative pyrosequencing technique. Although this study was based on an established murine cell line, the in vivo results suggest that the passage through the bloodstream may modulate methylation of selected genes in CTCs.

5. Outlook and Perspectives

The analysis of CTCs may provide relevant clinical and biological information on systemic cancer. Whereas phenotypic and genetic profiling have thus far constituted the main objectives of molecular CTC analysis, their epigenetic characterization has been a largely unexplored field. However, the existing data strongly encourages deeper epigenetic analyses of CTCs to address molecular mechanisms of metastasis, and to eventually improve diagnostic, monitor, and therapeutic tools as illustrated in Figure 2.
Considering the high potential of DNA methylation as a biomarker for the diagnosis, prognosis and monitoring of tumors, the low number of studies focusing on methylation analysis of CTCs is surprising. To date, no study of DNA methylation on pure isolated CTCs has been published. The most likely explanation for this state lies in the technical limitations associated with studying DNA methylation on few or single cells, which is necessary to investigate isolated CTCs. Hence, developing more robust protocols for systematic interrogation of DNA methylation on low numbers of cells should be a priority for the field.
Survival and fate of CTCs seem to be closely connected with phenotypical plasticity and the balance between epithelial- and mesenchymal-like states during EMT and MET [69]. Interestingly, existing data largely support the idea that remodeling of DNA methylation is required for EMT and has an impact on various steps of the metastatic cascade [141]. Therefore, DNA methylation is likely to play a key role in CTC survival and regulation of their metastatic potential. In this respect, it seems particularly promising to explore the dynamics of DNA methylation in CTCs and to elucidate to what extent it modulates their phenotypic plasticity. Another interesting aspect that could be studied on CTCs is the DNA methylation response to the different environmental cues [142,143] to which cancer cells are exposed during their journey to distant sites. In particular, it will be important to understand to what extent the blood environment (e.g., plasma composition, interaction with hematogenous cells) may modulate the epigenetic configuration of CTCs and thereby influence their fate. Not least, therapies, especially those by epigenetic drugs like azacytidine, will likely affect the DNA methylation patterns of CTCs. The potential use of CTCs to monitor the efficacy of such therapies should be explored.
Figure 2. Potential clinical and research applications of epigenetic analyses of CTCs. Standard blood samples or diagnostic leukapheresis products obtained from patients with cancer can be used for CTC enrichment, detection and isolation by different workflows. Isolated CTCs may be analyzed for DNA methylation by different technologies. The information collected from these analyses can be used to address basic molecular mechanisms of metastasis and, in the future, may also prove to be of value for clinical applications.
Figure 2. Potential clinical and research applications of epigenetic analyses of CTCs. Standard blood samples or diagnostic leukapheresis products obtained from patients with cancer can be used for CTC enrichment, detection and isolation by different workflows. Isolated CTCs may be analyzed for DNA methylation by different technologies. The information collected from these analyses can be used to address basic molecular mechanisms of metastasis and, in the future, may also prove to be of value for clinical applications.
Genes 06 01053 g002
In addition to DNA methylation, other epigenetic mechanisms cooperate in global and local regulation of gene transcription (e.g., histone modifications and chromatin accessibility). The study of these epigenetic layers in CTCs will certainly provide additional relevant information on the mechanisms of metastasis, but will require further technology development.
Considering the already reported phenotypic and genetic heterogeneity of CTCs, it appears likely that CTCs constitute an epigenetically heterogeneous population of cells. In this respect, it will be of great importance to explore whether any epigenetic signature can define sub-populations of CTCs with special clinical relevance, allowing, for example, to identify patients that can benefit from specific therapies. In fact, having overcome the initial technical challenge of detecting rare CTCs, the CTC field is now faced with the challenge of dissecting the heterogeneity of CTCs and identifying those among them with the highest clinical and diagnostic utility. Epigenetics could provide a substantial contribution to that aim. However, the proper characterization of their epigenetic status will require a systematic analysis of multiple CTCs at the single-cell level. So far, none of the methods available for analysis of DNA methylation of single cells has been used to analyze isolated CTCs and it is therefore difficult to estimate the performance of these methods in single CTCs. The results obtained from studies in other systems, especially on embryonic development, raise the promise that the technical barriers might soon be overcome and that it will be possible to comprehensively interrogate the epigenome of CTCs in the near future.

Acknowledgments

This work was supported in the laboratory of Constantin F. Pixberg, Rui P.L. Neves and Nikolas H. Stoecklein by the Düsseldorf School of Oncology (funded by the Comprehensive Cancer Center Düsseldorf/Deutsche Krebshilfe and the Medical Faculty HHU Düsseldorf), the Deutsche Krebshilfe (Project #109600/110429), and the Innovative Medicines Initiative Joint Undertaking (IMIJU) (Project CANCER-ID).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Wirtz, D.; Konstantopoulos, K.; Searson, P.C. The physics of cancer: The role of physical interactions and mechanical forces in metastasis. Nat. Rev. Cancer 2011, 11, 512–522. [Google Scholar]
  2. Greaves, M.; Maley, C.C. Clonal evolution in cancer. Nature 2012, 481, 306–313. [Google Scholar] [CrossRef] [PubMed]
  3. Pantel, K.; Speicher, M.R. The biology of circulating tumor cells. Oncogene 2015. [Google Scholar] [CrossRef] [PubMed]
  4. Mego, M.; Mani, S.A.; Cristofanilli, M. Molecular mechanisms of metastasis in breast cancer—Clinical applications. Nat. Rev. Clin. Oncol. 2010, 7, 693–701. [Google Scholar] [CrossRef] [PubMed]
  5. Schwarzenbach, H.; Hoon, D.S.; Pantel, K. Cell-free nucleic acids as biomarkers in cancer patients. Nat. Rev. Cancer 2011, 11, 426–437. [Google Scholar] [CrossRef] [PubMed]
  6. Fleischhacker, M.; Schmidt, B. Circulating nucleic acids (CNAS) and cancer—A survey. Biochim. Biophys. Acta 2007, 1775, 181–232. [Google Scholar] [CrossRef] [PubMed]
  7. Allard, W.J.; Matera, J.; Miller, M.C.; Repollet, M.; Connelly, M.C.; Rao, C.; Tibbe, A.G.; Uhr, J.W.; Terstappen, L.W. Tumor cells circulate in the peripheral blood of all major carcinomas but not in healthy subjects or patients with nonmalignant diseases. Clin. Cancer Res. 2004, 10, 6897–6904. [Google Scholar] [CrossRef] [PubMed]
  8. Cristofanilli, M.; Budd, G.T.; Ellis, M.J.; Stopeck, A.; Matera, J.; Miller, M.C.; Reuben, J.M.; Doyle, G.V.; Allard, W.J.; Terstappen, L.W.; et al. Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N. Engl. J. Med. 2004, 351, 781–791. [Google Scholar] [CrossRef] [PubMed]
  9. Hayes, D.F.; Cristofanilli, M.; Budd, G.T.; Ellis, M.J.; Stopeck, A.; Miller, M.C.; Matera, J.; Allard, W.J.; Doyle, G.V.; Terstappen, L.W. Circulating tumor cells at each follow-up time point during therapy of metastatic breast cancer patients predict progression-free and overall survival. Clin. Cancer Res. 2006, 12, 4218–4224. [Google Scholar] [CrossRef] [PubMed]
  10. Nole, F.; Munzone, E.; Zorzino, L.; Minchella, I.; Salvatici, M.; Botteri, E.; Medici, M.; Verri, E.; Adamoli, L.; Rotmensz, N.; et al. Variation of circulating tumor cell levels during treatment of metastatic breast cancer: Prognostic and therapeutic implications. Ann. Oncol. 2008, 19, 891–897. [Google Scholar] [CrossRef] [PubMed]
  11. Bidard, F.C.; Peeters, D.J.; Fehm, T.; Nole, F.; Gisbert-Criado, R.; Mavroudis, D.; Grisanti, S.; Generali, D.; Garcia-Saenz, J.A.; Stebbing, J.; et al. Clinical validity of circulating tumour cells in patients with metastatic breast cancer: A pooled analysis of individual patient data. Lancet Oncol. 2014, 15, 406–414. [Google Scholar] [CrossRef]
  12. Gruber, I.; Landenberger, N.; Staebler, A.; Hahn, M.; Wallwiener, D.; Fehm, T. Relationship between circulating tumor cells and peripheral T-cells in patients with primary breast cancer. Anticancer Res. 2013, 33, 2233–2238. [Google Scholar] [PubMed]
  13. Moreno, J.G.; Miller, M.C.; Gross, S.; Allard, W.J.; Gomella, L.G.; Terstappen, L.W. Circulating tumor cells predict survival in patients with metastatic prostate cancer. Urology 2005, 65, 713–718. [Google Scholar] [CrossRef] [PubMed]
  14. Cohen, S.J.; Punt, C.J.; Iannotti, N.; Saidman, B.H.; Sabbath, K.D.; Gabrail, N.Y.; Picus, J.; Morse, M.; Mitchell, E.; Miller, M.C.; et al. Relationship of circulating tumor cells to tumor response, progression-free survival, and overall survival in patients with metastatic colorectal cancer. J. Clin. Oncol. 2008, 26, 3213–3221. [Google Scholar] [CrossRef] [PubMed]
  15. De Bono, J.S.; Scher, H.I.; Montgomery, R.B.; Parker, C.; Miller, M.C.; Tissing, H.; Doyle, G.V.; Terstappen, L.W.; Pienta, K.J.; Raghavan, D. Circulating tumor cells predict survival benefit from treatment in metastatic castration-resistant prostate cancer. Clin. Cancer Res. 2008, 14, 6302–6309. [Google Scholar] [CrossRef] [PubMed]
  16. Serrano, M.J.; Rovira, P.S.; Martinez-Zubiaurre, I.; Rodriguez, M.D.; Fernandez, M.; Lorente, J.A. Dynamics of circulating tumor cells in early breast cancer under neoadjuvant therapy. Exp. Ther. Med. 2012, 4, 43–48. [Google Scholar] [PubMed]
  17. Pierga, J.Y.; Bidard, F.C.; Mathiot, C.; Brain, E.; Delaloge, S.; Giachetti, S.; de Cremoux, P.; Salmon, R.; Vincent-Salomon, A.; Marty, M. Circulating tumor cell detection predicts early metastatic relapse after neoadjuvant chemotherapy in large operable and locally advanced breast cancer in a phase II randomized trial. Clin. Cancer Res. 2008, 14, 7004–7010. [Google Scholar] [CrossRef] [PubMed]
  18. Rack, B.; Schindlbeck, C.; Juckstock, J.; Andergassen, U.; Hepp, P.; Zwingers, T.; Friedl, T.W.; Lorenz, R.; Tesch, H.; Fasching, P.A.; et al. Circulating tumor cells predict survival in early average-to-high risk breast cancer patients. J. Natl. Cancer Inst. 2014. [Google Scholar] [CrossRef] [PubMed]
  19. Smith, B.M.; Slade, M.J.; English, J.; Graham, H.; Luchtenborg, M.; Sinnett, H.D.; Cross, N.C.; Coombes, R.C. Response of circulating tumor cells to systemic therapy in patients with metastatic breast cancer: Comparison of quantitative polymerase chain reaction and immunocytochemical techniques. J. Clin. Oncol. 2000, 18, 1432–1439. [Google Scholar] [PubMed]
  20. Liu, M.C.; Shields, P.G.; Warren, R.D.; Cohen, P.; Wilkinson, M.; Ottaviano, Y.L.; Rao, S.B.; Eng-Wong, J.; Seillier-Moiseiwitsch, F.; Noone, A.M.; et al. Circulating tumor cells: A useful predictor of treatment efficacy in metastatic breast cancer. J. Clin. Oncol. 2009, 27, 5153–5159. [Google Scholar] [CrossRef] [PubMed]
  21. Smerage, J.B.; Barlow, W.E.; Hortobagyi, G.N.; Winer, E.P.; Leyland-Jones, B.; Srkalovic, G.; Tejwani, S.; Schott, A.F.; O’Rourke, M.A.; Lew, D.L.; et al. Circulating tumor cells and response to chemotherapy in metastatic breast cancer: SWOG S0500. J. Clin. Oncol. 2014, 32, 3483–3489. [Google Scholar] [CrossRef] [PubMed]
  22. Scher, H.I.; Heller, G.; Molina, A.; Attard, G.; Danila, D.C.; Jia, X.; Peng, W.; Sandhu, S.K.; Olmos, D.; Riisnaes, R.; et al. Circulating tumor cell biomarker panel as an individual-level surrogate for survival in metastatic castration-resistant prostate cancer. J. Clin. Oncol. 2015, 33, 1348–1355. [Google Scholar] [CrossRef] [PubMed]
  23. Prentice, R.L. Surrogate endpoints in clinical trials: Definition and operational criteria. Stat. Med. 1989, 8, 431–440. [Google Scholar] [CrossRef] [PubMed]
  24. Meng, S.; Tripathy, D.; Shete, S.; Ashfaq, R.; Haley, B.; Perkins, S.; Beitsch, P.; Khan, A.; Euhus, D.; Osborne, C.; et al. HER-2 gene amplification can be acquired as breast cancer progresses. Proc. Natl. Acad. Sci. USA 2004, 101, 9393–9398. [Google Scholar] [CrossRef] [PubMed]
  25. Pestrin, M.; Bessi, S.; Galardi, F.; Truglia, M.; Biggeri, A.; Biagioni, C.; Cappadona, S.; Biganzoli, L.; Giannini, A.; Di, L.A. Correlation of HER2 status between primary tumors and corresponding circulating tumor cells in advanced breast cancer patients. Breast Cancer Res. Treat. 2009, 118, 523–530. [Google Scholar] [CrossRef] [PubMed]
  26. Fehm, T.; Muller, V.; Aktas, B.; Janni, W.; Schneeweiss, A.; Stickeler, E.; Lattrich, C.; Lohberg, C.R.; Solomayer, E.; Rack, B.; et al. HER2 status of circulating tumor cells in patients with metastatic breast cancer: A prospective, multicenter trial. Breast Cancer Res. Treat. 2010, 124, 403–412. [Google Scholar] [CrossRef] [PubMed]
  27. Powell, A.A.; Talasaz, A.H.; Zhang, H.; Coram, M.A.; Reddy, A.; Deng, G.; Telli, M.L.; Advani, R.H.; Carlson, R.W.; Mollick, J.A.; et al. Single cell profiling of circulating tumor cells: Transcriptional heterogeneity and diversity from breast cancer cell lines. PLoS ONE 2012, 7, e33788. [Google Scholar] [CrossRef] [PubMed]
  28. Strati, A.; Markou, A.; Parisi, C.; Politaki, E.; Mavroudis, D.; Georgoulias, V.; Lianidou, E. Gene expression profile of circulating tumor cells in breast cancer by RT-qPCR. BMC Cancer 2011. [Google Scholar] [CrossRef] [PubMed]
  29. Antonarakis, E.S.; Lu, C.; Wang, H.; Luber, B.; Nakazawa, M.; Roeser, J.C.; Chen, Y.; Mohammad, T.A.; Chen, Y.; Fedor, H.L.; et al. AR-V7 and resistance to enzalutamide and abiraterone in prostate cancer. N. Engl. J. Med. 2014, 371, 1028–1038. [Google Scholar] [CrossRef] [PubMed]
  30. Hodgkinson, C.L.; Morrow, C.J.; Li, Y.; Metcalf, R.L.; Rothwell, D.G.; Trapani, F.; Polanski, R.; Burt, D.J.; Simpson, K.L.; Morris, K.; et al. Tumorigenicity and genetic profiling of circulating tumor cells in small-cell lung cancer. Nat. Med. 2014, 20, 897–903. [Google Scholar] [CrossRef] [PubMed]
  31. Baccelli, I.; Schneeweiss, A.; Riethdorf, S.; Stenzinger, A.; Schillert, A.; Vogel, V.; Klein, C.; Saini, M.; Bauerle, T.; Wallwiener, M.; et al. Identification of a population of blood circulating tumor cells from breast cancer patients that initiates metastasis in a xenograft assay. Nat. Biotechnol. 2013, 31, 539–544. [Google Scholar] [CrossRef] [PubMed]
  32. Zhang, L.; Ridgway, L.D.; Wetzel, M.D.; Ngo, J.; Yin, W.; Kumar, D.; Goodman, J.C.; Groves, M.D.; Marchetti, D. The identification and characterization of breast cancer CTCs competent for brain metastasis. Sci. Transl. Med. 2013. [Google Scholar] [CrossRef] [PubMed]
  33. Miller, M.C.; Doyle, G.V.; Terstappen, L.W. Significance of circulating tumor cells detected by the cellsearch system in patients with metastatic breast colorectal and prostate cancer. J. Oncol. 2010. [Google Scholar] [CrossRef] [PubMed]
  34. Fischer, J.C.; Niederacher, D.; Topp, S.A.; Honisch, E.; Schumacher, S.; Schmitz, N.; Zacarias Fohrding, L.; Vay, C.; Hoffmann, I.; Kasprowicz, N.S.; et al. Diagnostic leukapheresis enables reliable detection of circulating tumor cells of nonmetastatic cancer patients. Proc. Natl. Acad. Sci. USA 2013, 110, 16580–16585. [Google Scholar] [CrossRef] [PubMed]
  35. Tseng, J.Y.; Yang, C.Y.; Liang, S.C.; Liu, R.S.; Jiang, J.K.; Lin, C.H. Dynamic changes in numbers and properties of circulating tumor cells and their potential applications. Cancers 2014, 6, 2369–2386. [Google Scholar] [CrossRef] [PubMed]
  36. Yap, T.A.; Lorente, D.; Omlin, A.; Olmos, D.; de Bono, J.S. Circulating tumor cells: A multifunctional biomarker. Clin. Cancer Res. 2014, 20, 2553–2568. [Google Scholar] [CrossRef] [PubMed]
  37. Polzer, B.; Medoro, G.; Pasch, S.; Fontana, F.; Zorzino, L.; Pestka, A.; Andergassen, U.; Meier-Stiegen, F.; Czyz, Z.T.; Alberter, B.; et al. Molecular profiling of single circulating tumor cells with diagnostic intention. EMBO Mol. Med. 2014, 6, 1371–1386. [Google Scholar] [CrossRef] [PubMed]
  38. Neves, R.P.L.; Raba, K.; Schmidt, O.; Honisch, E.; Meier-Stiegen, F.; Behrens, B.; Möhlendick, B.; Fehm, T.; Neubauer, H.; Klein, C.; et al. Genomic high resolution profiling of single CKpos/CD45neg flow-sorting purified circulating tumour cells from patients with metastatic breast cancer. Clin. Chem. 2014, 60, 1290–1297. [Google Scholar] [PubMed]
  39. Harb, W.; Fan, A.; Tran, T.; Danila, D.C.; Keys, D.; Schwartz, M.; Ionescu-Zanetti, C. Mutational analysis of circulating tumor cells using a novel microfluidic collection device and qPCR assay. Transl. Oncol. 2013, 6, 528–538. [Google Scholar] [CrossRef] [PubMed]
  40. Talasaz, A.H.; Powell, A.A.; Huber, D.E.; Berbee, J.G.; Roh, K.H.; Yu, W.; Xiao, W.; Davis, M.M.; Pease, R.F.; Mindrinos, M.N.; et al. Isolating highly enriched populations of circulating epithelial cells and other rare cells from blood using a magnetic sweeper device. Proc. Natl. Acad. Sci. USA 2009, 106, 3970–3975. [Google Scholar] [CrossRef] [PubMed]
  41. Karabacak, N.M.; Spuhler, P.S.; Fachin, F.; Lim, E.J.; Pai, V.; Ozkumur, E.; Martel, J.M.; Kojic, N.; Smith, K.; Chen, P.I.; et al. Microfluidic, marker-free isolation of circulating tumor cells from blood samples. Nat. Protoc. 2014, 9, 694–710. [Google Scholar] [CrossRef] [PubMed]
  42. Saucedo-Zeni, N.; Mewes, S.; Niestroj, R.; Gasiorowski, L.; Murawa, D.; Nowaczyk, P.; Tomasi, T.; Weber, E.; Dworacki, G.; Morgenthaler, N.G.; et al. A novel method for the in vivo isolation of circulating tumor cells from peripheral blood of cancer patients using a functionalized and structured medical wire. Int. J. Oncol. 2012, 41, 1241–1250. [Google Scholar] [PubMed]
  43. Rao, C.G.; Chianese, D.; Doyle, G.V.; Miller, M.C.; Russell, T.; Sanders, R.A., Jr.; Terstappen, L.W. Expression of epithelial cell adhesion molecule in carcinoma cells present in blood and primary and metastatic tumors. Int. J. Oncol. 2005, 27, 49–57. [Google Scholar] [CrossRef] [PubMed]
  44. Gorges, T.M.; Tinhofer, I.; Drosch, M.; Rose, L.; Zollner, T.M.; Krahn, T.; von Ahsen, O. Circulating tumour cells escape from EpCAM-based detection due to epithelial-to-mesenchymal transition. BMC Cancer 2012. [Google Scholar] [CrossRef] [PubMed]
  45. Gires, O.; Stoecklein, N.H. Dynamic EpCAM expression on circulating and disseminating tumor cells: Causes and consequences. Cell. Mol. Life Sci. 2014, 71, 4393–4402. [Google Scholar] [CrossRef] [PubMed]
  46. Raimondi, C.; Gradilone, A.; Naso, G.; Vincenzi, B.; Petracca, A.; Nicolazzo, C.; Palazzo, A.; Saltarelli, R.; Spremberg, F.; Cortesi, E.; et al. Epithelial-mesenchymal transition and stemness features in circulating tumor cells from breast cancer patients. Breast Cancer Res. Treat. 2011, 130, 449–455. [Google Scholar] [CrossRef] [PubMed]
  47. Wu, S.; Liu, S.; Liu, Z.; Huang, J.; Pu, X.; Li, J.; Yang, D.; Deng, H.; Yang, N.; Xu, J. Classification of circulating tumor cells by epithelial-mesenchymal transition markers. PLoS ONE 2015, 10, e0123976. [Google Scholar] [CrossRef] [PubMed]
  48. Yu, M.; Bardia, A.; Wittner, B.S.; Stott, S.L.; Smas, M.E.; Ting, D.T.; Isakoff, S.J.; Ciciliano, J.C.; Wells, M.N.; Shah, A.M.; et al. Circulating breast tumor cells exhibit dynamic changes in epithelial and mesenchymal composition. Science 2013, 339, 580–584. [Google Scholar] [CrossRef] [PubMed]
  49. Barriere, G.; Fici, P.; Gallerani, G.; Fabbri, F.; Zoli, W.; Rigaud, M. Circulating tumor cells and epithelial, mesenchymal and stemness markers: Characterization of cell subpopulations. Ann. Transl. Med. 2014, 2. [Google Scholar] [CrossRef]
  50. Sieuwerts, A.M.; Kraan, J.; Bolt, J.; van der Spoel, P.; Elstrodt, F.; Schutte, M.; Martens, J.W.; Gratama, J.W.; Sleijfer, S.; Foekens, J.A. Anti-epithelial cell adhesion molecule antibodies and the detection of circulating normal-like breast tumor cells. J. Natl. Cancer Inst. 2009, 101, 61–66. [Google Scholar] [CrossRef] [PubMed]
  51. Low, W.S.; Wan Abas, W.A. Benchtop technologies for circulating tumor cells separation based on biophysical properties. BioMed Res. Int. 2015. [Google Scholar] [CrossRef] [PubMed]
  52. Vona, G.; Sabile, A.; Louha, M.; Sitruk, V.; Romana, S.; Schutze, K.; Capron, F.; Franco, D.; Pazzagli, M.; Vekemans, M.; et al. Isolation by size of epithelial tumor cells: A new method for the immunomorphological and molecular characterization of circulatingtumor cells. Am. J. Pathol. 2000, 156, 57–63. [Google Scholar] [CrossRef]
  53. Desitter, I.; Guerrouahen, B.S.; Benali-Furet, N.; Wechsler, J.; Janne, P.A.; Kuang, Y.; Yanagita, M.; Wang, L.; Berkowitz, J.A.; Distel, R.J.; et al. A new device for rapid isolation by size and characterization of rare circulating tumor cells. Anticancer Res. 2011, 31, 427–441. [Google Scholar] [PubMed]
  54. Adams, D.L.; Zhu, P.; Makarova, O.V.; Martin, S.S.; Charpentier, M.; Chumsri, S.; Li, S.; Amstutz, P.; Tang, C.M. The systematic study of circulating tumor cell isolation using lithographic microfilters. RSC Adv. 2014, 9, 4334–4342. [Google Scholar] [CrossRef] [PubMed]
  55. Fan, T.; Zhao, Q.; Chen, J.J.; Chen, W.T.; Pearl, M.L. Clinical significance of circulating tumor cells detected by an invasion assay in peripheral blood of patients with ovarian cancer. Gynecol. Oncol. 2009, 112, 185–191. [Google Scholar] [CrossRef] [PubMed]
  56. Goldberg, A.D.; Allis, C.D.; Bernstein, E. Epigenetics: A landscape takes shape. Cell 2007, 128, 635–638. [Google Scholar] [CrossRef] [PubMed]
  57. Jones, P.A. Functions of DNA methylation: Islands, start sites, gene bodies and beyond. Nat. Rev. Genet. 2012, 13, 484–492. [Google Scholar] [CrossRef] [PubMed]
  58. Maunakea, A.K.; Nagarajan, R.P.; Bilenky, M.; Ballinger, T.J.; D’Souza, C.; Fouse, S.D.; Johnson, B.E.; Hong, C.; Nielsen, C.; Zhao, Y.; et al. Conserved role of intragenic DNA methylation in regulating alternative promoters. Nature 2010, 466, 253–257. [Google Scholar] [CrossRef] [PubMed]
  59. Rauch, T.A.; Wu, X.; Zhong, X.; Riggs, A.D.; Pfeifer, G.P. A human B cell methylome at 100-base pair resolution. Proc. Natl. Acad. Sci. USA 2009, 106, 671–678. [Google Scholar] [CrossRef] [PubMed]
  60. Ehrlich, M. DNA hypomethylation in cancer cells. Epigenomics 2009, 1, 239–259. [Google Scholar] [CrossRef] [PubMed]
  61. Egger, G.; Liang, G.; Aparicio, A.; Jones, P.A. Epigenetics in human disease and prospects for epigenetic therapy. Nature 2004, 429, 457–463. [Google Scholar] [CrossRef] [PubMed]
  62. Schulz, W.A.D.; Dokun, O.Y. DNA methylation and human diseases: An overview. In DNA and RNA Modification Enzymes: Structure, Mechanism, Function and Evolution; Grosjean, H., Ed.; Landes Bioscience: Austin, TX, USA, 2009; pp. 103–116. [Google Scholar]
  63. Wilson, A.S.; Power, B.E.; Molloy, P.L. DNA hypomethylation and human diseases. Biochim. Biophys. Acta 2007, 1775, 138–162. [Google Scholar] [CrossRef] [PubMed]
  64. Witte, T.; Plass, C.; Gerhauser, C. Pan-cancer patterns of DNA methylation. Genome Med. 2014. [Google Scholar] [CrossRef] [PubMed]
  65. Fernandez, A.F.; Assenov, Y.; Martin-Subero, J.I.; Balint, B.; Siebert, R.; Taniguchi, H.; Yamamoto, H.; Hidalgo, M.; Tan, A.C.; Galm, O.; et al. A DNA methylation fingerprint of 1628 human samples. Genome Res. 2012, 22, 407–419. [Google Scholar] [CrossRef] [PubMed]
  66. Carmona, F.J.; Davalos, V.; Vidal, E.; Gomez, A.; Heyn, H.; Hashimoto, Y.; Vizoso, M.; Martinez-Cardus, A.; Sayols, S.; Ferreira, H.J.; et al. A comprehensive DNA methylation profile of epithelial-to-mesenchymal transition. Cancer Res. 2014, 74, 5608–5619. [Google Scholar] [CrossRef] [PubMed]
  67. Thiery, J.P.; Acloque, H.; Huang, R.Y.; Nieto, M.A. Epithelial-mesenchymal transitions in development and disease. Cell 2009, 139, 871–890. [Google Scholar] [CrossRef] [PubMed]
  68. Mani, S.A.; Guo, W.; Liao, M.J.; Eaton, E.N.; Ayyanan, A.; Zhou, A.Y.; Brooks, M.; Reinhard, F.; Zhang, C.C.; Shipitsin, M.; et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell 2008, 133, 704–715. [Google Scholar] [CrossRef] [PubMed]
  69. Kang, Y.; Pantel, K. Tumor cell dissemination: Emerging biological insights from animal models and cancer patients. Cancer Cell 2013, 23, 573–581. [Google Scholar] [CrossRef] [PubMed]
  70. Vrba, L.; Jensen, T.J.; Garbe, J.C.; Heimark, R.L.; Cress, A.E.; Dickinson, S.; Stampfer, M.R.; Futscher, B.W. Role for DNA methylation in the regulation of miR-200c and miR-141 expression in normal and cancer cells. PLoS ONE 2010, 5, e8697. [Google Scholar] [CrossRef] [PubMed]
  71. Ke, X.S.; Qu, Y.; Cheng, Y.; Li, W.C.; Rotter, V.; Oyan, A.M.; Kalland, K.H. Global profiling of histone and DNA methylation reveals epigenetic-based regulation of gene expression during epithelial to mesenchymal transition in prostate cells. BMC Genomics 2010. [Google Scholar] [CrossRef] [PubMed]
  72. Ulirsch, J.; Fan, C.; Knafl, G.; Wu, M.J.; Coleman, B.; Perou, C.M.; Swift-Scanlan, T. Vimentin DNA methylation predicts survival in breast cancer. Breast Cancer Res. Treat. 2013, 137, 383–396. [Google Scholar] [CrossRef] [PubMed]
  73. Fackler, M.J.; McVeigh, M.; Evron, E.; Garrett, E.; Mehrotra, J.; Polyak, K.; Sukumar, S.; Argani, P. DNA methylation of RASSF1A, HIN-1, RAR-β, Cyclin D2 and Twist in in situ and invasive lobular breast carcinoma. Int. J. Cancer 2003, 107, 970–975. [Google Scholar] [CrossRef] [PubMed]
  74. Neves, R.; Scheel, C.; Weinhold, S.; Honisch, E.; Iwaniuk, K.M.; Trompeter, H.I.; Niederacher, D.; Wernet, P.; Santourlidis, S.; Uhrberg, M. Role of DNA methylation in miR-200c/141 cluster silencing in invasive breast cancer cells. BMC Res. Notes 2010. [Google Scholar] [CrossRef] [PubMed]
  75. Davalos, V.; Moutinho, C.; Villanueva, A.; Boque, R.; Silva, P.; Carneiro, F.; Esteller, M. Dynamic epigenetic regulation of the microRNA-200 family mediates epithelial and mesenchymal transitions in human tumorigenesis. Oncogene 2011, 31, 2062–2074. [Google Scholar] [CrossRef] [PubMed]
  76. Steeg, P.S.; Ouatas, T.; Halverson, D.; Palmieri, D.; Salerno, M. Metastasis suppressor genes: Basic biology and potential clinical use. Clin. Breast Cancer 2003, 4, 51–62. [Google Scholar] [CrossRef] [PubMed]
  77. Wong, S.Y.; Haack, H.; Kissil, J.L.; Barry, M.; Bronson, R.T.; Shen, S.S.; Whittaker, C.A.; Crowley, D.; Hynes, R.O. Protein 4.1 B suppresses prostate cancer progression and metastasis. Proc. Natl. Acad. Sci. USA 2007, 104, 12784–12789. [Google Scholar] [CrossRef] [PubMed]
  78. Schulz, W.A.; Alexa, A.; Jung, V.; Hader, C.; Hoffmann, M.J.; Yamanaka, M.; Fritzsche, S.; Wlazlinski, A.; Muller, M.; Lengauer, T.; et al. Factor interaction analysis for chromosome 8 and DNA methylation alterations highlights innate immune response suppression and cytoskeletal changes in prostate cancer. Mol. Cancer 2007. [Google Scholar] [CrossRef] [PubMed]
  79. Fenaux, P.; Mufti, G.J.; Hellstrom-Lindberg, E.; Santini, V.; Finelli, C.; Giagounidis, A.; Schoch, R.; Gattermann, N.; Sanz, G.; List, A.; et al. Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: A randomised, open-label, phase III study. Lancet Oncol. 2009, 10, 223–232. [Google Scholar] [CrossRef]
  80. Fenaux, P.; Gattermann, N.; Seymour, J.F.; Hellstrom-Lindberg, E.; Mufti, G.J.; Duehrsen, U.; Gore, S.D.; Ramos, F.; Beyne-Rauzy, O.; List, A.; et al. Prolonged survival with improved tolerability in higher-risk myelodysplastic syndromes: Azacitidine compared with low dose ARA-C. Br. J. Haematol. 2010, 149, 244–249. [Google Scholar] [CrossRef] [PubMed]
  81. Fenaux, P.; Mufti, G.J.; Hellstrom-Lindberg, E.; Santini, V.; Gattermann, N.; Germing, U.; Sanz, G.; List, A.F.; Gore, S.; Seymour, J.F.; et al. Azacitidine prolongs overall survival compared with conventional care regimens in elderly patients with low bone marrow blast count acute myeloid leukemia. J. Clin. Oncol. 2010, 28, 562–569. [Google Scholar] [CrossRef] [PubMed]
  82. Blum, W.; Garzon, R.; Klisovic, R.B.; Schwind, S.; Walker, A.; Geyer, S.; Liu, S.; Havelange, V.; Becker, H.; Schaaf, L.; et al. Clinical response and miR-29b predictive significance in older AML patients treated with a 10-day schedule of decitabine. Proc. Natl. Acad. Sci. USA 2010, 107, 7473–7478. [Google Scholar] [CrossRef] [PubMed]
  83. Cashen, A.F.; Schiller, G.J.; O’Donnell, M.R.; DiPersio, J.F. Multicenter, phase II study of decitabine for the first-line treatment of older patients with acute myeloid leukemia. J. Clin. Oncol. 2010, 28, 556–561. [Google Scholar] [CrossRef] [PubMed]
  84. Fang, F.; Balch, C.; Schilder, J.; Breen, T.; Zhang, S.; Shen, C.; Li, L.; Kulesavage, C.; Snyder, A.J.; Nephew, K.P.; et al. A phase 1 and pharmacodynamic study of decitabine in combination with carboplatin in patients with recurrent, platinum-resistant, epithelial ovarian cancer. Cancer 2010, 116, 4043–4053. [Google Scholar] [CrossRef] [PubMed]
  85. Glasspool, R.M.; Brown, R.; Gore, M.E.; Rustin, G.J.; McNeish, I.A.; Wilson, R.H.; Pledge, S.; Paul, J.; Mackean, M.; Hall, G.D.; et al. A randomised, phase II trial of the DNA-hypomethylating agent 5-aza-2'-deoxycytidine (decitabine) in combination with carboplatin vs carboplatin alone in patients with recurrent, partially platinum-sensitive ovarian cancer. Br. J. Cancer 2014, 110, 1923–1929. [Google Scholar] [CrossRef] [PubMed]
  86. Fan, H.; Lu, X.; Wang, X.; Liu, Y.; Guo, B.; Zhang, Y.; Zhang, W.; Nie, J.; Feng, K.; Chen, M.; et al. Low-dose decitabine-based chemoimmunotherapy for patients with refractory advanced solid tumors: A phase I/II report. J. Immunol. Res. 2014. [Google Scholar] [CrossRef] [PubMed]
  87. Gilbert, M.R.; Wang, M.; Aldape, K.D.; Stupp, R.; Hegi, M.E.; Jaeckle, K.A.; Armstrong, T.S.; Wefel, J.S.; Won, M.; Blumenthal, D.T.; et al. Dose-dense temozolomide for newly diagnosed glioblastoma: A randomized phase III clinical trial. J. Clin. Oncol. 2013, 31, 4085–4091. [Google Scholar] [CrossRef] [PubMed]
  88. Tuominen, R.; Jewell, R.; van den Oord, J.J.; Wolter, P.; Stierner, U.; Lindholm, C.; Hertzman Johansson, C.; Linden, D.; Johansson, H.; Frostvik Stolt, M.; et al. Mgmt promoter methylation is associated with temozolomide response and prolonged progression-free survival in disseminated cutaneous melanoma. Int. J. Cancer 2015, 136, 2844–2853. [Google Scholar] [CrossRef] [PubMed]
  89. Weller, M.; Tabatabai, G.; Kastner, B.; Felsberg, J.; Steinbach, J.P.; Wick, A.; Schnell, O.; Hau, P.; Herrlinger, U.; Sabel, M.C.; et al. MGMT promoter methylation is a strong prognostic biomarker for benefit from dose-intensified temozolomide rechallenge in progressive glioblastoma: The DIRECTOR trial. Clin. Cancer Res. 2015, 21, 2057–2064. [Google Scholar] [CrossRef] [PubMed]
  90. Amatu, A.; Sartore-Bianchi, A.; Moutinho, C.; Belotti, A.; Bencardino, K.; Chirico, G.; Cassingena, A.; Rusconi, F.; Esposito, A.; Nichelatti, M.; et al. Promoter CpG island hypermethylation of the DNA repair enzyme MGMT predicts clinical response to dacarbazine in a phase II study for metastatic colorectal cancer. Clin. Cancer Res. 2013, 19, 2265–2272. [Google Scholar] [CrossRef] [PubMed]
  91. Zhou, Y.F.; Xu, W.; Wang, X.; Sun, J.S.; Xiang, J.J.; Li, Z.S.; Zhang, X.F. Negative methylation status of vimentin predicts improved prognosis in pancreatic carcinoma. World J. Gastroenterol. 2014, 20, 13172–13177. [Google Scholar] [CrossRef] [PubMed]
  92. Lee, M.K.; Jeong, E.M.; Kim, J.H.; Rho, S.B.; Lee, E.J. Aberrant methylation of the VIM promoter in uterine cervical squamous cell carcinoma. Oncology 2014, 86, 359–368. [Google Scholar] [PubMed]
  93. Itzkowitz, S.; Brand, R.; Jandorf, L.; Durkee, K.; Millholland, J.; Rabeneck, L.; Schroy, P.C., 3rd; Sontag, S.; Johnson, D.; Markowitz, S.; et al. A simplified, noninvasive stool DNA test for colorectal cancer detection. Am. J. Gastroenterol. 2008, 103, 2862–2870. [Google Scholar] [CrossRef] [PubMed]
  94. Costa, V.L.; Henrique, R.; Danielsen, S.A.; Duarte-Pereira, S.; Eknaes, M.; Skotheim, R.I.; Rodrigues, A.; Magalhaes, J.S.; Oliveira, J.; Lothe, R.A.; et al. Three epigenetic biomarkers, GDF15, TMEFF2, and VIM, accurately predict bladder cancer from DNA-based analyses of urine samples. Clin. Cancer Res. 2010, 16, 5842–5851. [Google Scholar] [CrossRef] [PubMed]
  95. Reinert, T.; Borre, M.; Christiansen, A.; Hermann, G.G.; Orntoft, T.F.; Dyrskjot, L. Diagnosis of bladder cancer recurrence based on urinary levels of EOMES, HOXA9, POU4F2, TWIST1, VIM, and ZNF154 hypermethylation. PLoS ONE 2012, 7, e46297. [Google Scholar] [CrossRef] [PubMed]
  96. Schuebel, K.E.; Chen, W.; Cope, L.; Glockner, S.C.; Suzuki, H.; Yi, J.M.; Chan, T.A.; van Neste, L.; van Criekinge, W.; van den Bosch, S.; et al. Comparing the DNA hypermethylome with gene mutations in human colorectal cancer. PLoS Genet. 2007, 3, 1709–1723. [Google Scholar] [CrossRef] [PubMed]
  97. Shen, L.; Waterland, R.A. Methods of DNA methylation analysis. Curr. Opin. Clin. Nutr. Metab. Care 2007, 10, 576–581. [Google Scholar] [CrossRef] [PubMed]
  98. Laird, P.W. Principles and challenges of genomewide DNA methylation analysis. Nat. Rev. Genet. 2010, 11, 191–203. [Google Scholar] [CrossRef] [PubMed]
  99. Frommer, M.; McDonald, L.E.; Millar, D.S.; Collis, C.M.; Watt, F.; Grigg, G.W.; Molloy, P.L.; Paul, C.L. A genomic sequencing protocol that yields a positive display of 5-methylcytosine residues in individual DNA strands. Proc. Natl. Acad. Sci. USA 1992, 89, 1827–1831. [Google Scholar] [CrossRef] [PubMed]
  100. Clark, S.J.; Harrison, J.; Paul, C.L.; Frommer, M. High sensitivity mapping of methylated cytosines. Nucleic Acids Res. 1994, 22, 2990–2997. [Google Scholar] [PubMed]
  101. Uhlmann, K.; Brinckmann, A.; Toliat, M.R.; Ritter, H.; Nurnberg, P. Evaluation of a potential epigenetic biomarker by quantitative methyl-single nucleotide polymorphism analysis. Electrophoresis 2002, 23, 4072–4079. [Google Scholar] [CrossRef] [PubMed]
  102. Ehrich, M.; Nelson, M.R.; Stanssens, P.; Zabeau, M.; Liloglou, T.; Xinarianos, G.; Cantor, C.R.; Field, J.K.; van den Boom, D. Quantitative high-throughput analysis of DNA methylation patterns by base-specific cleavage and mass spectrometry. Proc. Natl. Acad. Sci. USA 2005, 102, 15785–15790. [Google Scholar] [CrossRef] [PubMed]
  103. Ehrich, M.; Turner, J.; Gibbs, P.; Lipton, L.; Giovanneti, M.; Cantor, C.; van den Boom, D. Cytosine methylation profiling of cancer cell lines. Proc. Natl. Acad. Sci. USA 2008, 105, 4844–4849. [Google Scholar] [CrossRef] [PubMed]
  104. Ball, M.P.; Li, J.B.; Gao, Y.; Lee, J.H.; LeProust, E.M.; Park, I.H.; Xie, B.; Daley, G.Q.; Church, G.M. Targeted and genome-scale strategies reveal gene-body methylation signatures in human cells. Nat. Biotechnol. 2009, 27, 361–368. [Google Scholar] [CrossRef] [PubMed]
  105. Deng, J.; Shoemaker, R.; Xie, B.; Gore, A.; LeProust, E.M.; Antosiewicz-Bourget, J.; Egli, D.; Maherali, N.; Park, I.H.; Yu, J.; et al. Targeted bisulfite sequencing reveals changes in DNA methylation associated with nuclear reprogramming. Nat. Biotechnol. 2009, 27, 353–360. [Google Scholar] [CrossRef] [PubMed]
  106. Diep, D.; Plongthongkum, N.; Gore, A.; Fung, H.L.; Shoemaker, R.; Zhang, K. Library-free methylation sequencing with bisulfite padlock probes. Nat. Methods 2012, 9, 270–272. [Google Scholar] [CrossRef] [PubMed]
  107. Lee, E.J.; Pei, L.; Srivastava, G.; Joshi, T.; Kushwaha, G.; Choi, J.H.; Robertson, K.D.; Wang, X.; Colbourne, J.K.; Zhang, L.; et al. Targeted bisulfite sequencing by solution hybrid selection and massively parallel sequencing. Nucleic Acids Res. 2011. [Google Scholar] [CrossRef] [PubMed]
  108. Wang, J.; Jiang, H.; Ji, G.; Gao, F.; Wu, M.; Sun, J.; Luo, H.; Wu, J.; Wu, R.; Zhang, X. High resolution profiling of human exon methylation by liquid hybridization capture-based bisulfite sequencing. BMC Genomics 2011. [Google Scholar] [CrossRef] [PubMed]
  109. Bibikova, M.; Lin, Z.; Zhou, L.; Chudin, E.; Garcia, E.W.; Wu, B.; Doucet, D.; Thomas, N.J.; Wang, Y.; Vollmer, E.; et al. High-throughput DNA methylation profiling using universal bead arrays. Genome Res. 2006, 16, 383–393. [Google Scholar] [CrossRef] [PubMed]
  110. Bibikova, M.; Barnes, B.; Tsan, C.; Ho, V.; Klotzle, B.; Le, J.M.; Delano, D.; Zhang, L.; Schroth, G.P.; Gunderson, K.L.; et al. High density DNA methylation array with single CpG site resolution. Genomics 2011, 98, 288–295. [Google Scholar] [CrossRef] [PubMed]
  111. Lister, R.; Pelizzola, M.; Dowen, R.H.; Hawkins, R.D.; Hon, G.; Tonti-Filippini, J.; Nery, J.R.; Lee, L.; Ye, Z.; Ngo, Q.M.; et al. Human DNA methylomes at base resolution show widespread epigenomic differences. Nature 2009, 462, 315–322. [Google Scholar] [CrossRef] [PubMed]
  112. Laurent, L.; Wong, E.; Li, G.; Huynh, T.; Tsirigos, A.; Ong, C.T.; Low, H.M.; Kin Sung, K.W.; Rigoutsos, I.; Loring, J.; et al. Dynamic changes in the human methylome during differentiation. Genome Res. 2010, 20, 320–331. [Google Scholar] [CrossRef] [PubMed]
  113. Meissner, A.; Gnirke, A.; Bell, G.W.; Ramsahoye, B.; Lander, E.S.; Jaenisch, R. Reduced representation bisulfite sequencing for comparative high-resolution DNA methylation analysis. Nucleic Acids Res. 2005, 33, 5868–5877. [Google Scholar] [CrossRef] [PubMed]
  114. Meissner, A.; Mikkelsen, T.S.; Gu, H.; Wernig, M.; Hanna, J.; Sivachenko, A.; Zhang, X.; Bernstein, B.E.; Nusbaum, C.; Jaffe, D.B.; et al. Genome-scale DNA methylation maps of pluripotent and differentiated cells. Nature 2008, 454, 766–770. [Google Scholar] [CrossRef] [PubMed]
  115. Guo, H.; Zhu, P.; Wu, X.; Li, X.; Wen, L.; Tang, F. Single-cell methylome landscapes of mouse embryonic stem cells and early embryos analyzed using reduced representation bisulfite sequencing. Genome Res. 2013, 23, 2126–2135. [Google Scholar] [CrossRef] [PubMed]
  116. Gravina, S.; Ganapathi, S.; Vijg, J. Single-cell, locus-specific bisulfite sequencing (SLBS) for direct detection of epimutations in DNA methylation patterns. Nucleic Acids Res. 2015. [Google Scholar] [CrossRef] [PubMed]
  117. Chimonidou, M.; Strati, A.; Tzitzira, A.; Sotiropoulou, G.; Malamos, N.; Georgoulias, V.; Lianidou, E.S. DNA methylation of tumor suppressor and metastasis suppressor genes in circulating tumor cells. Clin. Chem. 2011, 57, 1169–1177. [Google Scholar] [CrossRef] [PubMed]
  118. Chimonidou, M.; Strati, A.; Malamos, N.; Georgoulias, V.; Lianidou, E.S. SOX17 promoter methylation in circulating tumor cells and matched cell-free DNA isolated from plasma of patients with breast cancer. Clin. Chem. 2013, 59, 270–279. [Google Scholar] [CrossRef] [PubMed]
  119. Chimonidou, M.; Kallergi, G.; Georgoulias, V.; Welch, D.R.; Lianidou, E.S. Breast cancer metastasis suppressor-1 promoter methylation in primary breast tumors and corresponding circulating tumor cells. Mol. Cancer Res. 2013, 11, 1248–1257. [Google Scholar] [CrossRef] [PubMed]
  120. Ni, J.; Abrahamson, M.; Zhang, M.; Fernandez, M.A.; Grubb, A.; Su, J.; Yu, G.L.; Li, Y.; Parmelee, D.; Xing, L.; et al. Cystatin E is a novel human cysteine proteinase inhibitor with structural resemblance to family 2 cystatins. J. Biol. Chem. 1997, 272, 10853–10858. [Google Scholar] [PubMed]
  121. Sotiropoulou, G.; Anisowicz, A.; Sager, R. Identification, cloning, and characterization of cystatin M, a novel cysteine proteinase inhibitor, down-regulated in breast cancer. J. Biol. Chem. 1997, 272, 903–910. [Google Scholar] [CrossRef] [PubMed]
  122. Zhang, J.; Shridhar, R.; Dai, Q.; Song, J.; Barlow, S.C.; Yin, L.; Sloane, B.F.; Miller, F.R.; Meschonat, C.; Li, B.D.; et al. Cystatin M: A novel candidate tumor suppressor gene for breast cancer. Cancer Res. 2004, 64, 6957–6964. [Google Scholar] [CrossRef] [PubMed]
  123. Ai, L.; Kim, W.J.; Kim, T.Y.; Fields, C.R.; Massoll, N.A.; Robertson, K.D.; Brown, K.D. Epigenetic silencing of the tumor suppressor cystatin M occurs during breast cancer progression. Cancer Res. 2006, 66, 7899–7909. [Google Scholar] [CrossRef] [PubMed]
  124. Kioulafa, M.; Balkouranidou, I.; Sotiropoulou, G.; Kaklamanis, L.; Mavroudis, D.; Georgoulias, V.; Lianidou, E.S. Methylation of cystatin M promoter is associated with unfavorable prognosis in operable breast cancer. Int. J. Cancer 2009, 125, 2887–2892. [Google Scholar] [CrossRef] [PubMed]
  125. Seraj, M.J.; Samant, R.S.; Verderame, M.F.; Welch, D.R. Functional evidence for a novel human breast carcinoma metastasis suppressor, BRMS1, encoded at chromosome 11q13. Cancer Res. 2000, 60, 2764–2769. [Google Scholar] [PubMed]
  126. Metge, B.J.; Frost, A.R.; King, J.A.; Dyess, D.L.; Welch, D.R.; Samant, R.S.; Shevde, L.A. Epigenetic silencing contributes to the loss of BRMS1 expression in breast cancer. Clin. Exp. Metastasis 2008, 25, 753–763. [Google Scholar] [CrossRef] [PubMed]
  127. Zhang, Z.; Yamashita, H.; Toyama, T.; Yamamoto, Y.; Kawasoe, T.; Iwase, H. Reduced expression of the breast cancer metastasis suppressor 1 mRNA is correlated with poor progress in breast cancer. Clin. Cancer Res. 2006, 12, 6410–6414. [Google Scholar] [CrossRef] [PubMed]
  128. Stark, A.M.; Tongers, K.; Maass, N.; Mehdorn, H.M.; Held-Feindt, J. Reduced metastasis-suppressor gene mRNA-expression in breast cancer brain metastases. J. Cancer Res. Clin. Oncol. 2005, 131, 191–198. [Google Scholar] [CrossRef] [PubMed]
  129. Hicks, D.G.; Yoder, B.J.; Short, S.; Tarr, S.; Prescott, N.; Crowe, J.P.; Dawson, A.E.; Budd, G.T.; Sizemore, S.; Cicek, M.; et al. Loss of breast cancer metastasis suppressor 1 protein expression predicts reduced disease-free survival in subsets of breast cancer patients. Clin. Cancer Res. 2006, 12, 6702–6708. [Google Scholar] [CrossRef] [PubMed]
  130. Zhang, W.; Glockner, S.C.; Guo, M.; Machida, E.O.; Wang, D.H.; Easwaran, H.; Van Neste, L.; Herman, J.G.; Schuebel, K.E.; Watkins, D.N.; et al. Epigenetic inactivation of the canonical Wnt antagonist SRY-box containing gene 17 in colorectal cancer. Cancer Res. 2008, 68, 2764–2772. [Google Scholar] [CrossRef] [PubMed]
  131. Sinner, D.; Kordich, J.J.; Spence, J.R.; Opoka, R.; Rankin, S.; Lin, S.C.; Jonatan, D.; Zorn, A.M.; Wells, J.M. SOX17 AND SOX4 differentially regulate beta-catenin/T-cell factor activity and proliferation of colon carcinoma cells. Mol. Cell. Biol. 2007, 27, 7802–7815. [Google Scholar] [CrossRef] [PubMed]
  132. Fu, Y.; Tan, H.S.; Wei, J.L.; Zhu, C.R.; Jiang, J.X.; Zhu, Y.X.; Cai, F.L.; Chong, M.H.; Ren, C.L. Decreased expression of SOX17 is associated with tumor progression and poor prognosis in breast cancer. Tumour Biol. 2015. [Google Scholar] [CrossRef] [PubMed]
  133. Lu, J.; Zhang, G.; Cheng, Y.; Tang, Y.; Dong, Z.; McElwee, K.J.; Li, G. Reduced expression of SRY-box containing gene 17 correlates with an unfavorable melanoma patient survival. Oncol. Rep. 2014, 32, 2571–2579. [Google Scholar] [CrossRef] [PubMed]
  134. Friedlander, T.W.; Ngo, V.T.; Dong, H.; Premasekharan, G.; Weinberg, V.; Doty, S.; Zhao, Q.; Gilbert, E.G.; Ryan, C.J.; Chen, W.T.; et al. Detection and characterization of invasive circulating tumor cells derived from men with metastatic castration-resistant prostate cancer. Int. J. Cancer 2014, 134, 2284–2293. [Google Scholar] [CrossRef] [PubMed]
  135. Ogunwobi, O.O.; Puszyk, W.; Dong, H.J.; Liu, C. Epigenetic upregulation of HGF and c-Met drives metastasis in hepatocellular carcinoma. PLoS ONE 2013, 8, e63765. [Google Scholar] [CrossRef] [PubMed]
  136. Li, L.; Choi, J.Y.; Lee, K.M.; Sung, H.; Park, S.K.; Oze, I.; Pan, K.F.; You, W.C.; Chen, Y.X.; Fang, J.Y.; et al. DNA methylation in peripheral blood: A potential biomarker for cancer molecular epidemiology. J. Epidemiol. 2012, 22, 384–394. [Google Scholar] [CrossRef] [PubMed]
  137. Lu, J.; Fan, T.; Zhao, Q.; Zeng, W.; Zaslavsky, E.; Chen, J.J.; Frohman, M.A.; Golightly, M.G.; Madajewicz, S.; Chen, W.T. Isolation of circulating epithelial and tumor progenitor cells with an invasive phenotype from breast cancer patients. Int. J. Cancer 2010, 126, 669–683. [Google Scholar] [CrossRef] [PubMed]
  138. Paris, P.L.; Kobayashi, Y.; Zhao, Q.; Zeng, W.; Sridharan, S.; Fan, T.; Adler, H.L.; Yera, E.R.; Zarrabi, M.H.; Zucker, S.; et al. Functional phenotyping and genotyping of circulating tumor cells from patients with castration resistant prostate cancer. Cancer Lett. 2009, 277, 164–173. [Google Scholar] [CrossRef] [PubMed]
  139. Friedlander, T.W.; Roy, R.; Tomlins, S.A.; Ngo, V.T.; Kobayashi, Y.; Azameera, A.; Rubin, M.A.; Pienta, K.J.; Chinnaiyan, A.; Ittmann, M.M.; et al. Common structural and epigenetic changes in the genome of castration-resistant prostate cancer. Cancer Res. 2012, 72, 616–625. [Google Scholar] [CrossRef] [PubMed]
  140. Trusolino, L.; Bertotti, A.; Comoglio, P.M. MET signalling: Principles and functions in development, organ regeneration and cancer. Nat. Rev. Mol. Cell Biol. 2010, 11, 834–848. [Google Scholar] [CrossRef] [PubMed]
  141. Tam, W.L.; Weinberg, R.A. The epigenetics of epithelial-mesenchymal plasticity in cancer. Nat. Med. 2013, 19, 1438–1449. [Google Scholar] [CrossRef] [PubMed]
  142. Jaenisch, R.; Bird, A. Epigenetic regulation of gene expression: How the genome integrates intrinsic and environmental signals. Nat. Genet. 2003, 33, 245–254. [Google Scholar] [CrossRef] [PubMed]
  143. DesRochers, T.M.; Shamis, Y.; Alt-Holland, A.; Kudo, Y.; Takata, T.; Wang, G.; Jackson-Grusby, L.; Garlick, J.A. The 3D tissue microenvironment modulates DNA methylation and E-cadherin expression in squamous cell carcinoma. Epigenetics 2012, 7, 34–46. [Google Scholar] [CrossRef] [PubMed]

Share and Cite

MDPI and ACS Style

Pixberg, C.F.; Schulz, W.A.; Stoecklein, N.H.; Neves, R.P.L. Characterization of DNA Methylation in Circulating Tumor Cells. Genes 2015, 6, 1053-1075. https://doi.org/10.3390/genes6041053

AMA Style

Pixberg CF, Schulz WA, Stoecklein NH, Neves RPL. Characterization of DNA Methylation in Circulating Tumor Cells. Genes. 2015; 6(4):1053-1075. https://doi.org/10.3390/genes6041053

Chicago/Turabian Style

Pixberg, Constantin F., Wolfgang A. Schulz, Nikolas H. Stoecklein, and Rui P. L. Neves. 2015. "Characterization of DNA Methylation in Circulating Tumor Cells" Genes 6, no. 4: 1053-1075. https://doi.org/10.3390/genes6041053

Article Metrics

Back to TopTop